1
|
Dreier JP, Joerk A, Uchikawa H, Horst V, Lemale CL, Radbruch H, McBride DW, Vajkoczy P, Schneider UC, Xu R. All Three Supersystems-Nervous, Vascular, and Immune-Contribute to the Cortical Infarcts After Subarachnoid Hemorrhage. Transl Stroke Res 2025; 16:96-118. [PMID: 38689162 PMCID: PMC11772491 DOI: 10.1007/s12975-024-01242-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 05/02/2024]
Abstract
The recently published DISCHARGE-1 trial supports the observations of earlier autopsy and neuroimaging studies that almost 70% of all focal brain damage after aneurysmal subarachnoid hemorrhage are anemic infarcts of the cortex, often also affecting the white matter immediately below. The infarcts are not limited by the usual vascular territories. About two-fifths of the ischemic damage occurs within ~ 48 h; the remaining three-fifths are delayed (within ~ 3 weeks). Using neuromonitoring technology in combination with longitudinal neuroimaging, the entire sequence of both early and delayed cortical infarct development after subarachnoid hemorrhage has recently been recorded in patients. Characteristically, cortical infarcts are caused by acute severe vasospastic events, so-called spreading ischemia, triggered by spontaneously occurring spreading depolarization. In locations where a spreading depolarization passes through, cerebral blood flow can drastically drop within a few seconds and remain suppressed for minutes or even hours, often followed by high-amplitude, sustained hyperemia. In spreading depolarization, neurons lead the event, and the other cells of the neurovascular unit (endothelium, vascular smooth muscle, pericytes, astrocytes, microglia, oligodendrocytes) follow. However, dysregulation in cells of all three supersystems-nervous, vascular, and immune-is very likely involved in the dysfunction of the neurovascular unit underlying spreading ischemia. It is assumed that subarachnoid blood, which lies directly on the cortex and enters the parenchyma via glymphatic channels, triggers these dysregulations. This review discusses the neuroglial, neurovascular, and neuroimmunological dysregulations in the context of spreading depolarization and spreading ischemia as critical elements in the pathogenesis of cortical infarcts after subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Jens P Dreier
- Center for Stroke Research Berlin, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- Department of Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany.
- Einstein Center for Neurosciences Berlin, Berlin, Germany.
| | - Alexander Joerk
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Hiroki Uchikawa
- Barrow Aneurysm & AVM Research Center, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Viktor Horst
- Center for Stroke Research Berlin, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Institute of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Coline L Lemale
- Center for Stroke Research Berlin, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Helena Radbruch
- Institute of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Devin W McBride
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ulf C Schneider
- Department of Neurosurgery, Cantonal Hospital of Lucerne and University of Lucerne, Lucerne, Switzerland
| | - Ran Xu
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- DZHK, German Centre for Cardiovascular Research, Berlin, Germany
| |
Collapse
|
2
|
Bijnen M, Sridhar S, Keller A, Greter M. Brain macrophages in vascular health and dysfunction. Trends Immunol 2025; 46:46-60. [PMID: 39732528 DOI: 10.1016/j.it.2024.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/08/2024] [Accepted: 11/19/2024] [Indexed: 12/30/2024]
Abstract
Diverse macrophage populations inhabit the rodent and human central nervous system (CNS), including microglia in the parenchyma and border-associated macrophages (BAMs) in the meninges, choroid plexus, and perivascular spaces. These innate immune phagocytes are essential in brain development and maintaining homeostasis, but they also play diverse roles in neurological diseases. In this review, we highlight the emerging roles of CNS macrophages in regulating vascular function in health and disease. We discuss that, in addition to microglia, BAMs, including perivascular macrophages, play roles in supporting vascular integrity and maintaining blood flow. We highlight recent advancements in understanding how these macrophages are implicated in protecting against vascular dysfunction and modulating the progression of cerebrovascular diseases, as seen in vessel-associated neurodegeneration.
Collapse
Affiliation(s)
- Mitchell Bijnen
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sucheta Sridhar
- Department of Neurosurgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Annika Keller
- Department of Neurosurgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
3
|
Pedard M, Prevost L, Carpena C, Holleran B, Desrues L, Dubois M, Nicola C, Gruel R, Godefroy D, Deffieux T, Tanter M, Ali C, Leduc R, Prézeau L, Gandolfo P, Morin F, Wurtz O, Bonnard T, Vivien D, Castel H. The urotensin II receptor triggers an early meningeal response and a delayed macrophage-dependent vasospasm after subarachnoid hemorrhage in male mice. Nat Commun 2024; 15:8430. [PMID: 39341842 PMCID: PMC11439053 DOI: 10.1038/s41467-024-52654-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
Subarachnoid hemorrhage (SAH) can be associated with neurological deficits and has profound consequences for mortality and morbidity. Cerebral vasospasm (CVS) and delayed cerebral ischemia affect neurological outcomes in SAH patients, but their mechanisms are not fully understood, and effective treatments are limited. Here, we report that urotensin II receptor UT plays a pivotal role in both early events and delayed mechanisms following SAH in male mice. Few days post-SAH, UT expression is triggered by blood or hemoglobin in the leptomeningeal compartment. UT contributes to perimeningeal glia limitans astrocyte reactivity, microvascular alterations and neuroinflammation independent of CNS-associated macrophages (CAMs). Later, CAM-dependent vascular inflammation and subsequent CVS develop, leading to cognitive dysfunction. In an SAH model using humanized UTh+/h+ male mice, we show that post-SAH CVS and behavioral deficits, mediated by UT through Gq/PLC/Ca2+ signaling, are prevented by UT antagonists. These results highlight the potential of targeting UT pathways to reduce early meningeal response and delayed cerebral ischemia in SAH patients.
Collapse
Affiliation(s)
- Martin Pedard
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Lucie Prevost
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Camille Carpena
- Institut de Génomique Fonctionnelle, Univ. Montpellier, CNRS, Inserm, Montpellier, France
| | - Brian Holleran
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Laurence Desrues
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Martine Dubois
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Celeste Nicola
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Roxane Gruel
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - David Godefroy
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
- Univ Rouen Normandie, Inserm, Normandie Univ, NorDiC UMR 1239, Rouen, France
| | - Thomas Deffieux
- Institute Physics for Medicine, Inserm U1273, CNRS UMR 8631, ESPCI Paris, Paris Sciences et Lettres PSL University, Paris, France
| | - Mickael Tanter
- Institute Physics for Medicine, Inserm U1273, CNRS UMR 8631, ESPCI Paris, Paris Sciences et Lettres PSL University, Paris, France
| | - Carine Ali
- Normandie Université, UNICAEN, INSERM U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Richard Leduc
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Laurent Prézeau
- Institut de Génomique Fonctionnelle, Univ. Montpellier, CNRS, Inserm, Montpellier, France
| | - Pierrick Gandolfo
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Fabrice Morin
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Olivier Wurtz
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Thomas Bonnard
- Normandie Université, UNICAEN, INSERM U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Denis Vivien
- Normandie Université, UNICAEN, INSERM U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
- Centre Hospitalier Universitaire Caen, Department of Clinical Research, Caen, France
| | - Hélène Castel
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France.
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France.
| |
Collapse
|
4
|
Pacholko A, Iadecola C. Hypertension, Neurodegeneration, and Cognitive Decline. Hypertension 2024; 81:991-1007. [PMID: 38426329 PMCID: PMC11023809 DOI: 10.1161/hypertensionaha.123.21356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Elevated blood pressure is a well-established risk factor for age-related cognitive decline. Long linked to cognitive impairment on vascular bases, increasing evidence suggests a potential association of hypertension with the neurodegenerative pathology underlying Alzheimer disease. Hypertension is well known to disrupt the structural and functional integrity of the cerebral vasculature. However, the mechanisms by which these alterations lead to brain damage, enhance Alzheimer pathology, and promote cognitive impairment remain to be established. Furthermore, critical questions concerning whether lowering blood pressure by antihypertensive medications prevents cognitive impairment have not been answered. Recent developments in neurovascular biology, brain imaging, and epidemiology, as well as new clinical trials, have provided insights into these critical issues. In particular, clinical and basic findings on the link between neurovascular dysfunction and the pathobiology of neurodegeneration have shed new light on the overlap between vascular and Alzheimer pathology. In this review, we will examine the progress made in the relationship between hypertension and cognitive impairment and, after a critical evaluation of the evidence, attempt to identify remaining knowledge gaps and future research directions that may advance our understanding of one of the leading health challenges of our time.
Collapse
Affiliation(s)
- Anthony Pacholko
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| |
Collapse
|
5
|
Uchikawa H, Uekawa K, Hasegawa Y. Perivascular macrophages in cerebrovascular diseases. Exp Neurol 2024; 374:114680. [PMID: 38185314 DOI: 10.1016/j.expneurol.2024.114680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/10/2023] [Accepted: 01/02/2024] [Indexed: 01/09/2024]
Abstract
Cerebrovascular diseases are a major cause of stroke and dementia, both requiring long-term care. These diseases involve multiple pathophysiologies, with mitochondrial dysfunction being a crucial contributor to the initiation of inflammation, apoptosis, and oxidative stress, resulting in injuries to neurovascular units that include neuronal cell death, endothelial cell death, glial activation, and blood-brain barrier disruption. To maintain brain homeostasis against these pathogenic conditions, brain immune cells, including border-associated macrophages and microglia, play significant roles as brain innate immunity cells in the pathophysiology of cerebrovascular injury. Although microglia have long been recognized as significant contributors to neuroinflammation, attention has recently shifted to border-associated macrophages, such as perivascular macrophages (PVMs), which have been studied based on their crucial roles in the brain. These cells are strategically positioned around the walls of brain vessels, where they mainly perform critical functions, such as perivascular drainage, cerebrovascular flexibility, phagocytic activity, antigen presentation, activation of inflammatory responses, and preservation of blood-brain barrier integrity. Although PVMs act as scavenger and surveillant cells under normal conditions, these cells exert harmful effects under pathological conditions. PVMs detect mitochondrial dysfunction in injured cells and implement pathological changes to regulate brain homeostasis. Therefore, PVMs are promising as they play a significant role in mitochondrial dysfunction and, in turn, disrupt the homeostatic condition. Herein, we summarize the significant roles of PVMs in cerebrovascular diseases, especially ischemic and hemorrhagic stroke and dementia, mainly in correlation with inflammation. A better understanding of the biology and pathobiology of PVMs may lead to new insights on and therapeutic strategies for cerebrovascular diseases.
Collapse
Affiliation(s)
- Hiroki Uchikawa
- Department of Translational Neuroscience, Barrow Aneurysm and AVM Research Center, Barrow Neurological Institute, Phoenix, AZ, USA; Department of Neurosurgery, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Kumamoto, Japan
| | - Ken Uekawa
- Department of Neurosurgery, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Kumamoto, Japan
| | - Yu Hasegawa
- Department of Pharmaceutical Science, School of Pharmacy at Fukuoka, International University of Health and Welfare, Okawa, Fukuoka, Japan.
| |
Collapse
|
6
|
Gupta S, Singh P, Sharma B. Montelukast Ameliorates 2K1C-Hypertension Induced Endothelial Dysfunction and Associated Vascular Dementia. Curr Hypertens Rev 2024; 20:23-35. [PMID: 38192137 DOI: 10.2174/0115734021276985231204092425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/31/2023] [Accepted: 11/13/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND Declined kidney function associated with hypertension is a danger for cognitive deficits, dementia, and brain injury. Cognitive decline and vascular dementia (VaD) are serious public health concerns, which highlights the urgent need for study on the risk factors for cognitive decline. Cysteinyl leukotriene (CysLT1) receptors are concerned with regulating cognition, motivation, inflammatory processes, and neurogenesis. OBJECTIVE This research aims to examine the consequence of montelukast (specific CysLT1 antagonist) in renovascular hypertension 2-kidney-1-clip-2K1C model-triggered VaD in experimental animals. METHODS 2K1C tactics were made to prompt renovascular hypertension in mature male rats. Morris water maze was employed to measure cognition. Mean arterial pressure (MAP), serum nitrite levels, aortic superoxide content, vascular endothelial activity, brain's oxidative stress (diminished glutathione, raised lipid peroxides), inflammatory markers (IL-10, IL-6, TNF-α), cholinergic activity (raised acetylcholinesterase), and cerebral injury (staining of 2, 3, 5- triphenylterazolium chloride) were also examined. RESULTS Montelukast in doses of 5.0 and 10.0 mg kg-1 was used intraperitoneally as the treatment drug. Along with cognitive deficits, 2K1C-operated rats showed elevated MAP, endothelial dysfunction, brain oxidative stress, inflammation, and cerebral damage with diminished serum nitrite/nitrate. Montelukast therapy significantly and dose-dependently mitigated the 2K1Chypertension- provoked impaired behaviors, biochemistry, endothelial functions, and cerebral infarction. CONCLUSION The 2K1C tactic caused renovascular hypertension and associated VaD, which was mitigated via targeted regulation of CysLT1 receptors by montelukast administration. Therefore, montelukast may be taken into consideration for the evaluation of its complete potential in renovascular-hypertension-induced VaD.
Collapse
MESH Headings
- Animals
- Sulfides
- Cyclopropanes
- Acetates/pharmacology
- Quinolines/pharmacology
- Male
- Dementia, Vascular/physiopathology
- Dementia, Vascular/drug therapy
- Dementia, Vascular/metabolism
- Dementia, Vascular/psychology
- Leukotriene Antagonists/pharmacology
- Oxidative Stress/drug effects
- Hypertension, Renovascular/physiopathology
- Hypertension, Renovascular/drug therapy
- Hypertension, Renovascular/metabolism
- Disease Models, Animal
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/physiopathology
- Endothelium, Vascular/metabolism
- Receptors, Leukotriene/metabolism
- Inflammation Mediators/metabolism
- Cognition/drug effects
- Rats, Wistar
- Brain/drug effects
- Brain/metabolism
- Brain/physiopathology
- Rats
- Maze Learning/drug effects
Collapse
Affiliation(s)
- Surbhi Gupta
- Department of Pharmacology, School of Pharmacy, Bharat Institute of Technology, Partapur Bypass, Meerut, Uttar Pradesh, India
| | - Prabhat Singh
- Faculty of Pharmacy, Swami Vivekanand Subharti University, Meerut, Uttar Pradesh, India
| | - Bhupesh Sharma
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University Uttar Pradesh, Noida, India
| |
Collapse
|
7
|
Sankowski R, Süß P, Benkendorff A, Böttcher C, Fernandez-Zapata C, Chhatbar C, Cahueau J, Monaco G, Gasull AD, Khavaran A, Grauvogel J, Scheiwe C, Shah MJ, Heiland DH, Schnell O, Markfeld-Erol F, Kunze M, Zeiser R, Priller J, Prinz M. Multiomic spatial landscape of innate immune cells at human central nervous system borders. Nat Med 2024; 30:186-198. [PMID: 38123840 PMCID: PMC10803260 DOI: 10.1038/s41591-023-02673-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/30/2023] [Indexed: 12/23/2023]
Abstract
The innate immune compartment of the human central nervous system (CNS) is highly diverse and includes several immune-cell populations such as macrophages that are frequent in the brain parenchyma (microglia) and less numerous at the brain interfaces as CNS-associated macrophages (CAMs). Due to their scantiness and particular location, little is known about the presence of temporally and spatially restricted CAM subclasses during development, health and perturbation. Here we combined single-cell RNA sequencing, time-of-flight mass cytometry and single-cell spatial transcriptomics with fate mapping and advanced immunohistochemistry to comprehensively characterize the immune system at human CNS interfaces with over 356,000 analyzed transcriptomes from 102 individuals. We also provide a comprehensive analysis of resident and engrafted myeloid cells in the brains of 15 individuals with peripheral blood stem cell transplantation, revealing compartment-specific engraftment rates across different CNS interfaces. Integrated multiomic and high-resolution spatial transcriptome analysis of anatomically dissected glioblastoma samples shows regionally distinct myeloid cell-type distributions driven by hypoxia. Notably, the glioblastoma-associated hypoxia response was distinct from the physiological hypoxia response in fetal microglia and CAMs. Our results highlight myeloid diversity at the interfaces of the human CNS with the periphery and provide insights into the complexities of the human brain's immune system.
Collapse
Affiliation(s)
- Roman Sankowski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Patrick Süß
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Molecular Neurology, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, Erlangen, Germany
| | - Alexander Benkendorff
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Chotima Böttcher
- Neuropsychiatry Unit and Laboratory of Molecular Psychiatry, Charité, Universitätsmedizin Berlin and DZNE, Berlin, Germany
| | - Camila Fernandez-Zapata
- Neuropsychiatry Unit and Laboratory of Molecular Psychiatry, Charité, Universitätsmedizin Berlin and DZNE, Berlin, Germany
| | - Chintan Chhatbar
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jonathan Cahueau
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gianni Monaco
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg, Germany
| | - Adrià Dalmau Gasull
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ashkan Khavaran
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jürgen Grauvogel
- Department of Neurosurgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Scheiwe
- Department of Neurosurgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Mukesch Johannes Shah
- Department of Neurosurgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dieter Henrik Heiland
- Department of Neurosurgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Schnell
- Department of Neurosurgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Filiz Markfeld-Erol
- Department of Gynecology, Obstetrics, and Perinatology, Faculty of Medicine, University Hospital, Freiburg, Germany
| | - Mirjam Kunze
- Department of Gynecology, Obstetrics, and Perinatology, Faculty of Medicine, University Hospital, Freiburg, Germany
| | - Robert Zeiser
- Department of Internal Medicine I, Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Josef Priller
- Neuropsychiatry Unit and Laboratory of Molecular Psychiatry, Charité, Universitätsmedizin Berlin and DZNE, Berlin, Germany
- Department of Psychiatry and Psychotherapy, School of Medicine and Health, Technical University of Munich, Munich, Germany
- University of Edinburgh and UK DRI, Edinburgh, UK
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
8
|
Silvin A, Qian J, Ginhoux F. Brain macrophage development, diversity and dysregulation in health and disease. Cell Mol Immunol 2023; 20:1277-1289. [PMID: 37365324 PMCID: PMC10616292 DOI: 10.1038/s41423-023-01053-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
Brain macrophages include microglia in the parenchyma, border-associated macrophages in the meningeal-choroid plexus-perivascular space, and monocyte-derived macrophages that infiltrate the brain under various disease conditions. The vast heterogeneity of these cells has been elucidated over the last decade using revolutionary multiomics technologies. As such, we can now start to define these various macrophage populations according to their ontogeny and their diverse functional programs during brain development, homeostasis and disease pathogenesis. In this review, we first outline the critical roles played by brain macrophages during development and healthy aging. We then discuss how brain macrophages might undergo reprogramming and contribute to neurodegenerative disorders, autoimmune diseases, and glioma. Finally, we speculate about the most recent and ongoing discoveries that are prompting translational attempts to leverage brain macrophages as prognostic markers or therapeutic targets for diseases that affect the brain.
Collapse
Affiliation(s)
- Aymeric Silvin
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif, 94800, France
| | - Jiawen Qian
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Florent Ginhoux
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif, 94800, France.
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, 138648, Republic of Singapore.
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, 169856, Singapore.
| |
Collapse
|
9
|
Dennis MR, Pires PW, Banek CT. Vascular Dysfunction in Polycystic Kidney Disease: A Mini-Review. J Vasc Res 2023; 60:125-136. [PMID: 37536302 PMCID: PMC10947982 DOI: 10.1159/000531647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 06/10/2023] [Indexed: 08/05/2023] Open
Abstract
Polycystic kidney disease (PKD) is one of the most common hereditary kidney diseases, which is characterized by progressive cyst growth and secondary hypertension. In addition to cystogenesis and renal abnormalities, patients with PKD can develop vascular abnormalities and cardiovascular complications. Progressive cyst growth substantially alters renal structure and culminates into end-stage renal disease. There remains no cure beyond renal transplantation, and treatment options remain largely limited to chronic renal replacement therapy. In addition to end-stage renal disease, patients with PKD also present with hypertension and cardiovascular disease, yet the timing and interactions between the cardiovascular and renal effects of PKD progression are understudied. Here, we review the vascular dysfunction found in clinical and preclinical models of PKD, including the clinical manifestations and relationship to hypertension, stroke, and related cardiovascular diseases. Finally, our discussion also highlights the critical questions and emerging areas in vascular research in PKD.
Collapse
Affiliation(s)
- Melissa R Dennis
- Department of Physiology, University of Arizona Health Sciences Center, Tucson, Arizona, USA
| | - Paulo W Pires
- Department of Physiology, University of Arizona Health Sciences Center, Tucson, Arizona, USA
| | - Christopher T Banek
- Department of Physiology, University of Arizona Health Sciences Center, Tucson, Arizona, USA
| |
Collapse
|
10
|
Uchikawa H, Kameno K, Kai K, Kajiwara S, Fujimori K, Uekawa K, Fujiwara Y, Mukasa A, Kim-Mitsuyama S, Hasegawa Y. Pretreatment with Clodronate Improved Neurological Function by Preventing Reduction of Posthemorrhagic Cerebral Blood Flow in Experimental Subarachnoid Hemorrhage. Neurocrit Care 2023; 39:207-217. [PMID: 37308726 DOI: 10.1007/s12028-023-01754-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 05/08/2023] [Indexed: 06/14/2023]
Abstract
BACKGROUND Brain perivascular macrophages (PVMs) are potential treatment targets for subarachnoid hemorrhage (SAH), and previous studies revealed that their depletion by clodronate (CLD) improved outcomes after experimental SAH. However, the underlying mechanisms are not well understood. Therefore, we investigated whether reducing PVMs by CLD pretreatment improves SAH prognosis by inhibiting posthemorrhagic impairment of cerebral blood flow (CBF). METHODS In total, 80 male Sprague-Dawley rats received an intracerebroventricular injection of the vehicle (liposomes) or CLD. Subsequently, the rats were categorized into the prechiasmatic saline injection (sham) and blood injection (SAH) groups after 72 h. We assessed its effects on weak and severe SAH, which were induced by 200- and 300-µL arterial blood injections, respectively. In addition, neurological function at 72 h and CBF changes from before the intervention to 5 min after were assessed in rats after sham/SAH induction as the primary and secondary end points, respectively. RESULTS CLD significantly reduced PVMs before SAH induction. Although pretreatment with CLD in the weak SAH group provided no additive effects on the primary end point, rats in the severe SAH group showed significant improvement in the rotarod test. In the severe SAH group, CLD inhibited acute reduction of CBF and tended to decrease hypoxia-inducible factor 1α expression. Furthermore, CLD reduced the number of PVMs in rats subjected to sham and SAH surgery, although no effects were observed in oxidative stress and inflammation. CONCLUSIONS Our study proposes that pretreatment with CLD-targeting PVMs can improve the prognosis of severe SAH through a candidate mechanism of inhibition of posthemorrhagic CBF reduction.
Collapse
Affiliation(s)
- Hiroki Uchikawa
- Department of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
- Department of Neurosurgery, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Koki Kameno
- Department of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
- Department of Neurosurgery, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Keitaro Kai
- Department of Neurosurgery, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Sosho Kajiwara
- Department of Neurosurgery, Kurume University School of Medicine, Kurume, Japan
| | - Kana Fujimori
- Department of Neurosurgery, Kurume University School of Medicine, Kurume, Japan
| | - Ken Uekawa
- Department of Neurosurgery, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Yukio Fujiwara
- Department of Cell Pathology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Akitake Mukasa
- Department of Neurosurgery, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Shokei Kim-Mitsuyama
- Department of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Yu Hasegawa
- Department of Neurosurgery, Kurume University School of Medicine, Kurume, Japan.
- Department of Pharmaceutical Science, School of Pharmacy at Fukuoka, International University of Health and Welfare, Okawa, Japan.
| |
Collapse
|
11
|
Chen S, Li J, Meng S, He T, Shi Z, Wang C, Wang Y, Cao H, Huang Y, Zhang Y, Gong Y, Gao Y. Microglia and macrophages in the neuro-glia-vascular unit: From identity to functions. Neurobiol Dis 2023; 179:106066. [PMID: 36889483 DOI: 10.1016/j.nbd.2023.106066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Although both are myeloid cells located surrounding cerebral vasculature, vessel-associated microglia (VAM) and perivascular macrophages (PVMs) can be distinguished by their distinct morphologies, signatures and microscopic location. As key component of neuro-glia-vascular unit (NGVU), they play prominent roles in neurovasculature development and pathological process of various central nervous system (CNS) diseases, including phagocytosis, angiogenesis, vessel damage/protection and blood flow regulation, therefore serving as potential targets for therapeutics of a broad array of CNS diseases. Herein, we will provide a comprehensive overview of heterogeneity of VAM/PVMs, highlight limitations of current understanding in this field, and discuss possible directions of future investigations.
Collapse
Affiliation(s)
- Shuning Chen
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jiaying Li
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Shan Meng
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Tingyu He
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ziyu Shi
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Chenran Wang
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yana Wang
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Hui Cao
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yichen Huang
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yue Zhang
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ye Gong
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China.
| | - Yanqin Gao
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
12
|
Drieu A, Du S, Storck SE, Rustenhoven J, Papadopoulos Z, Dykstra T, Zhong F, Kim K, Blackburn S, Mamuladze T, Harari O, Karch CM, Bateman RJ, Perrin R, Farlow M, Chhatwal J, Hu S, Randolph GJ, Smirnov I, Kipnis J. Parenchymal border macrophages regulate the flow dynamics of the cerebrospinal fluid. Nature 2022; 611:585-593. [PMID: 36352225 PMCID: PMC9899827 DOI: 10.1038/s41586-022-05397-3] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 09/29/2022] [Indexed: 11/11/2022]
Abstract
Macrophages are important players in the maintenance of tissue homeostasis1. Perivascular and leptomeningeal macrophages reside near the central nervous system (CNS) parenchyma2, and their role in CNS physiology has not been sufficiently well studied. Given their continuous interaction with the cerebrospinal fluid (CSF) and strategic positioning, we refer to these cells collectively as parenchymal border macrophages (PBMs). Here we demonstrate that PBMs regulate CSF flow dynamics. We identify a subpopulation of PBMs that express high levels of CD163 and LYVE1 (scavenger receptor proteins), closely associated with the brain arterial tree, and show that LYVE1+ PBMs regulate arterial motion that drives CSF flow. Pharmacological or genetic depletion of PBMs led to accumulation of extracellular matrix proteins, obstructing CSF access to perivascular spaces and impairing CNS perfusion and clearance. Ageing-associated alterations in PBMs and impairment of CSF dynamics were restored after intracisternal injection of macrophage colony-stimulating factor. Single-nucleus RNA sequencing data obtained from patients with Alzheimer's disease (AD) and from non-AD individuals point to changes in phagocytosis, endocytosis and interferon-γ signalling on PBMs, pathways that are corroborated in a mouse model of AD. Collectively, our results identify PBMs as new cellular regulators of CSF flow dynamics, which could be targeted pharmacologically to alleviate brain clearance deficits associated with ageing and AD.
Collapse
Affiliation(s)
- Antoine Drieu
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA.
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA.
| | - Siling Du
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Immunology Graduate Program, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Steffen E Storck
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Justin Rustenhoven
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Zachary Papadopoulos
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Immunology Graduate Program, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Taitea Dykstra
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Fenghe Zhong
- Department of Biomedical Engineering, Danforth Campus, Washington University in St Louis, St Louis, MO, USA
| | - Kyungdeok Kim
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Susan Blackburn
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Tornike Mamuladze
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Immunology Graduate Program, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Oscar Harari
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Celeste M Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Randall J Bateman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Richard Perrin
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | | | - Jasmeer Chhatwal
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Song Hu
- Department of Biomedical Engineering, Danforth Campus, Washington University in St Louis, St Louis, MO, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Igor Smirnov
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA.
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA.
- Immunology Graduate Program, School of Medicine, Washington University in St Louis, St Louis, MO, USA.
| |
Collapse
|
13
|
Chambers LC, Diaz-Otero JM, Fisher CL, Jackson WF, Dorrance AM. Mineralocorticoid receptor antagonism improves transient receptor potential vanilloid 4-dependent dilation of cerebral parenchymal arterioles and cognition in a genetic model of hypertension. J Hypertens 2022; 40:1722-1734. [PMID: 35943101 PMCID: PMC9373385 DOI: 10.1097/hjh.0000000000003208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
OBJECTIVE In a model of secondary hypertension, mineralocorticoid receptor (MR) antagonism during the development of hypertension prevents the impairment of transient receptor potential vanilloid 4 (TRPV4) activation in parenchymal arterioles (PAs) and cognitive impairment. However, it is unknown whether MR antagonism can improve these impairments when treatment begins after the onset of essential hypertension. We tested the hypothesis that MR activation in stroke-prone spontaneously hypertensive rats (SHRSP) leads to impaired TRPV4-mediated dilation in PAs that is associated with cognitive dysfunction and neuroinflammation. METHODS 20-22-week-old male SHRSP ± eplerenone (EPL; 100 mg/kg daily for 4 weeks) were compared to normotensive Sprague-Dawley (SD) rats. Pressure myography was used to assess PA function. Cognition was tested using Y-maze. Neuroinflammation was assessed using immunofluorescence and qRT-PCR. RESULTS Carbachol-mediated endothelium-dependent dilation was impaired in SHRSP, and MR antagonism improved this without affecting myogenic tone. Dilation to TRPV4 agonist GSK1016790A was impaired in SHRSP, and ELP treatment restored this. Intermediate conductance potassium channel (IKCa)/small conductance potassium channel (SKCa)-mediated dilation was impaired by hypertension and unaffected by EPL treatment. TRPV4 and IKCa/SKCa channel mRNA expression were reduced in PAs from hypertensive rats, and EPL did not improve this. Impairments in PA dilation in SHRSP were associated with cognitive decline, microglial activation, reactive astrogliosis, and neuroinflammation; cognitive and inflammatory changes were improved with MR blockade. CONCLUSIONS These data advance our understanding of the effects of hypertension on cerebral arterioles using a clinically relevant model and treatment paradigm. Our studies suggest TRPV4 and the MR are potential therapeutic targets to improve cerebrovascular function and cognition during hypertension.
Collapse
Affiliation(s)
- Laura C Chambers
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, USA
| | | | | | | | | |
Collapse
|
14
|
Specification of CNS macrophage subsets occurs postnatally in defined niches. Nature 2022; 604:740-748. [PMID: 35444273 DOI: 10.1038/s41586-022-04596-2] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/28/2022] [Indexed: 02/08/2023]
Abstract
All tissue-resident macrophages of the central nervous system (CNS)-including parenchymal microglia, as well as CNS-associated macrophages (CAMs1) such as meningeal and perivascular macrophages2-7-are part of the CNS endogenous innate immune system that acts as the first line of defence during infections or trauma2,8-10. It has been suggested that microglia and all subsets of CAMs are derived from prenatal cellular sources in the yolk sac that were defined as early erythromyeloid progenitors11-15. However, the precise ontogenetic relationships, the underlying transcriptional programs and the molecular signals that drive the development of distinct CAM subsets in situ are poorly understood. Here we show, using fate-mapping systems, single-cell profiling and cell-specific mutants, that only meningeal macrophages and microglia share a common prenatal progenitor. By contrast, perivascular macrophages originate from perinatal meningeal macrophages only after birth in an integrin-dependent manner. The establishment of perivascular macrophages critically requires the presence of arterial vascular smooth muscle cells. Together, our data reveal a precisely timed process in distinct anatomical niches for the establishment of macrophage subsets in the CNS.
Collapse
|
15
|
Warrington JP, Shao Q, Clayton AM, Maeda KJ, Beckett AG, Garrett MR, Sasser JM. Pial Vessel-Associated Microglia/Macrophages Increase in Female Dahl-SS/Jr Rats Independent of Pregnancy History. Int J Mol Sci 2022; 23:3384. [PMID: 35328808 PMCID: PMC8950577 DOI: 10.3390/ijms23063384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/18/2022] [Accepted: 03/18/2022] [Indexed: 11/21/2022] Open
Abstract
As the resident immune cells of the central nervous system, microglia have a wide range of functions such as surveillance, phagocytosis, and signaling through production of chemokines and cytokines. Recent studies have identified and characterized macrophages residing at the meninges, a series of layers surrounding the brain and spinal cord. While perivascular microglia within the brain parenchyma increase following chronic hypertension, there are no reports of changes at the meninges, and specifically, associated with the pial vasculature. Thus, we used female Sprague Dawley and Dahl salt-sensitive (SS/Jr) rat brains, stained for ionized calcium-binding adapter molecule (Iba1), and characterized microglia/macrophages associated with pial vessels in the posterior brain. Results indicate that Iba1+ pial vessel-associated microglia (PVAM) completely surrounded the vessels in brains from the Dahl-SS/Jr rats. PVAM density was significantly higher and distance between PVAMs lower in Dahl-SS/Jr compared to the Sprague Dawley rat brains. Pregnancy history did not affect these findings. While the functional role of these cells are not known, we contextualize our novel findings with that of other studies assessing or characterizing myeloid cells at the borders of the CNS (meninges and choroid plexus) and perivascular macrophages and propose their possible origin in the Dahl-SS/Jr model of chronic hypertension.
Collapse
Affiliation(s)
- Junie P. Warrington
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (Q.S.); (A.M.C.); (A.G.B.)
- Department of Neurobiology & Anatomical Sciences, Neuro Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Qingmei Shao
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (Q.S.); (A.M.C.); (A.G.B.)
| | - Ahsia M. Clayton
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (Q.S.); (A.M.C.); (A.G.B.)
| | - Kenji J. Maeda
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (K.J.M.); (M.R.G.); (J.M.S.)
| | - Ashtin G. Beckett
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (Q.S.); (A.M.C.); (A.G.B.)
| | - Michael R. Garrett
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (K.J.M.); (M.R.G.); (J.M.S.)
| | - Jennifer M. Sasser
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (K.J.M.); (M.R.G.); (J.M.S.)
| |
Collapse
|
16
|
Wenstedt EFE, van Croonenburg TJ, van den Born BJH, Van den Bossche J, Hooijmans CR, Vogt L. The effect of macrophage-targeted interventions on blood pressure - a systematic review and meta-analysis of preclinical studies. Transl Res 2021; 230:123-138. [PMID: 33166696 DOI: 10.1016/j.trsl.2020.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/08/2020] [Accepted: 11/03/2020] [Indexed: 01/01/2023]
Abstract
An increasing body of evidence shows a role for macrophages and monocytes (as their precursors) in hypertension, but with conflicting results with regard to whether they are protective or harmful. Therefore, we systematically reviewed the effect of macrophage interventions on blood pressure in animal models, to explore which factors determine the blood pressure increasing vs. decreasing effect. A search in PubMED and EMBASE yielded 9620 records, 26 of which were included. Eighteen studies (involving 22 different experiments (k = 22)) performed macrophage depletion, whereas 12 studies specifically deleted certain macrophage proteins. The blood pressure effects of macrophage depletion were highly various and directed toward both directions, as expected, which could not be reduced to differences in animal species or methods of hypertension induction. Prespecified subgroup analysis did reveal a potential role for the route in which the macrophage-depleting agent is being administrated (intraperitoneal vs intravenous subgroup difference of P = 0.07 (k = 22), or P < 0.001 in studies achieving considerable (ie, >50%) depletion (k = 18)). Along with findings from specific macrophage protein deletion studies-showing that deletion of one single macrophage protein (like TonEBP, endothelin-B, EP4, NOX-2 and the angiotensin II type 1 receptor) can alter blood pressure responses to hypertensive stimuli-the indication that each route has its specific depletion pattern regarding targeted tissues and macrophage phenotypes suggests a determinative role for these features. These hypothesis-generating results encourage more detailed depletion characterization of each technique by direct experimental comparisons, providing a chance to obtain more knowledge on which macrophages are beneficial versus detrimental in hypertension development.
Collapse
Affiliation(s)
- Eliane F E Wenstedt
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Thirza J van Croonenburg
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Bert-Jan H van den Born
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Jan Van den Bossche
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Carlijn R Hooijmans
- Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE), Department of Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Liffert Vogt
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| |
Collapse
|
17
|
Pagano PJ, Cifuentes-Pagano E. The Enigmatic Vascular NOX: From Artifact to Double Agent of Change: Arthur C. Corcoran Memorial Lecture - 2019. Hypertension 2021; 77:275-283. [PMID: 33390049 DOI: 10.1161/hypertensionaha.120.13897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
NOXs (NADPH oxidases) comprise a family of proteins whose primary function is the production of reactive oxygen species, namely, superoxide anion and hydrogen peroxide. The prototype first being discovered and characterized in neutrophils, multiple NOXs are now known to be broadly expressed in cell and organ systems and whose phylogeny spans countless life forms beginning with prokaryotes. This long-enduring evolutionary conservation underscores the importance of fundamental NOX functions. This review chronicles a personal perspective of the field beginning with the discovery of NOXs in the vasculature and the advances achieved through the years as to our understanding of their mechanisms of action and role in oxidative stress and disease. Furthermore, applications of isoform-selective inhibitors to dissect the role of NOX isozymes in vascular biology, focusing on inflammation, pulmonary hypertension, and aging are described.
Collapse
Affiliation(s)
- Patrick J Pagano
- Department of Pharmacology and Chemical Biology, Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA
| | - Eugenia Cifuentes-Pagano
- Department of Pharmacology and Chemical Biology, Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA
| |
Collapse
|
18
|
Key Enzymes for the Mevalonate Pathway in the Cardiovascular System. J Cardiovasc Pharmacol 2021; 77:142-152. [PMID: 33538531 DOI: 10.1097/fjc.0000000000000952] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/15/2020] [Indexed: 11/25/2022]
Abstract
ABSTRACT Isoprenylation is an important post-transcriptional modification of small GTPases required for their activation and function. Isoprenoids, including farnesyl pyrophosphate (FPP) and geranylgeranyl pyrophosphate, are indispensable for isoprenylation by serving as donors of a prenyl moiety to small G proteins. In the human body, isoprenoids are mainly generated by the mevalonate pathway (also known as the cholesterol-synthesis pathway). The hydroxymethylglutaryl coenzyme A reductase catalyzes the first rate-limiting steps of the mevalonate pathway, and its inhibitor (statins) are widely used as lipid-lowering agents. In addition, the FPP synthase is also of critical importance for the regulation of the isoprenoids production, for which the inhibitor is mainly used in the treatment of osteoporosis. Synthetic FPP can be further used to generate geranylgeranyl pyrophosphate and cholesterol. Recent studies suggest a role for isoprenoids in the genesis and development of cardiovascular disorders, such as pathological cardiac hypertrophy, fibrosis, endothelial dysfunction, and fibrotic responses of smooth-muscle cells. Furthermore, statins and FPP synthase inhibitors have also been applied for the management of heart failure and other cardiovascular diseases rather than their clinical use for hyperlipidemia or bone diseases. In this review, we focus on the function of several critical enzymes, including hydroxymethylglutaryl coenzyme A reductase, FPP synthase, farnesyltransferase, and geranylgeranyltransferase in the mevalonate pathway which are involved in regulating the generation of isoprenoids and isoprenylation of small GTPases, and their pathophysiological role in the cardiovascular system. Moreover, we summarize recent research into applications of statins and the FPP synthase inhibitors to treat cardiovascular diseases, rather than for their traditional indications respectively.
Collapse
|
19
|
Freitas-Andrade M, Raman-Nair J, Lacoste B. Structural and Functional Remodeling of the Brain Vasculature Following Stroke. Front Physiol 2020; 11:948. [PMID: 32848875 PMCID: PMC7433746 DOI: 10.3389/fphys.2020.00948] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Maintenance of cerebral blood vessel integrity and regulation of cerebral blood flow ensure proper brain function. The adult human brain represents only a small portion of the body mass, yet about a quarter of the cardiac output is dedicated to energy consumption by brain cells at rest. Due to a low capacity to store energy, brain health is heavily reliant on a steady supply of oxygen and nutrients from the bloodstream, and is thus particularly vulnerable to stroke. Stroke is a leading cause of disability and mortality worldwide. By transiently or permanently limiting tissue perfusion, stroke alters vascular integrity and function, compromising brain homeostasis and leading to widespread consequences from early-onset motor deficits to long-term cognitive decline. While numerous lines of investigation have been undertaken to develop new pharmacological therapies for stroke, only few advances have been made and most clinical trials have failed. Overall, our understanding of the acute and chronic vascular responses to stroke is insufficient, yet a better comprehension of cerebrovascular remodeling following stroke is an essential prerequisite for developing novel therapeutic options. In this review, we present a comprehensive update on post-stroke cerebrovascular remodeling, an important and growing field in neuroscience, by discussing cellular and molecular mechanisms involved, sex differences, limitations of preclinical research design and future directions.
Collapse
Affiliation(s)
| | - Joanna Raman-Nair
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
20
|
Kierdorf K, Masuda T, Jordão MJC, Prinz M. Macrophages at CNS interfaces: ontogeny and function in health and disease. Nat Rev Neurosci 2019; 20:547-562. [PMID: 31358892 DOI: 10.1038/s41583-019-0201-x] [Citation(s) in RCA: 241] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2019] [Indexed: 12/16/2022]
Abstract
The segregation and limited regenerative capacity of the CNS necessitate a specialized and tightly regulated resident immune system that continuously guards the CNS against invading pathogens and injury. Immunity in the CNS has generally been attributed to neuron-associated microglia in the parenchyma, whose origin and functions have recently been elucidated. However, there are several other specialized macrophage populations at the CNS borders, including dural, leptomeningeal, perivascular and choroid plexus macrophages (collectively known as CNS-associated macrophages (CAMs)), whose origins and roles in health and disease have remained largely uncharted. CAMs are thought to be involved in regulating the fine balance between the proper segregation of the CNS, on the one hand, and the essential exchange between the CNS parenchyma and the periphery, on the other. Recent studies that have been empowered by major technological advances have shed new light on these cells and suggest central roles for CAMs in CNS physiology and in the pathogenesis of diseases.
Collapse
Affiliation(s)
- Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Takahiro Masuda
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany. .,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Centre for Biological Signalling Studies (BIOSS), University of Freiburg, Freiburg, Germany.
| |
Collapse
|
21
|
Abstract
Hypertension has emerged as a leading cause of age-related cognitive impairment. Long known to be associated with dementia caused by vascular factors, hypertension has more recently been linked also to Alzheimer disease-the major cause of dementia in older people. Thus, although midlife hypertension is a risk factor for late-life dementia, hypertension may also promote the neurodegenerative pathology underlying Alzheimer disease. The mechanistic bases of these harmful effects remain to be established. Hypertension is well known to alter in the structure and function of cerebral blood vessels, but how these cerebrovascular effects lead to cognitive impairment and promote Alzheimer disease pathology is not well understood. Furthermore, critical questions also concern whether treatment of hypertension prevents cognitive impairment, the blood pressure threshold for treatment, and the antihypertensive agents to be used. Recent advances in neurovascular biology, epidemiology, brain imaging, and biomarker development have started to provide new insights into these critical issues. In this review, we will examine the progress made to date, and, after a critical evaluation of the evidence, we will highlight questions still outstanding and seek to provide a path forward for future studies.
Collapse
Affiliation(s)
- Costantino Iadecola
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York (C.I.)
| | - Rebecca F Gottesman
- Departments of Neurology (R.F.G.), Johns Hopkins University, Baltimore, MD
- Epidemiology (R.F.G.), Johns Hopkins University, Baltimore, MD
| |
Collapse
|
22
|
Santisteban MM, Iadecola C. Hypertension, dietary salt and cognitive impairment. J Cereb Blood Flow Metab 2018; 38:2112-2128. [PMID: 30295560 PMCID: PMC6282225 DOI: 10.1177/0271678x18803374] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/31/2018] [Indexed: 12/12/2022]
Abstract
Dementia is growing at an alarming rate worldwide. Although Alzheimer disease is the leading cause, over 50% of individuals diagnosed with Alzheimer disease have vascular lesions at autopsy. There has been an increasing appreciation of the pathogenic role of vascular risk factors in cognitive impairment caused by neurodegeneration. Midlife hypertension is a leading risk factor for late-life dementia. Hypertension alters cerebrovascular structure, impairs the major factors regulating the cerebral microcirculation, and promotes Alzheimer pathology. Experimental studies have identified brain perivascular macrophages as the major free radical source mediating neurovascular dysfunction of hypertension. Recent evidence indicates that high dietary salt may also induce cognitive impairment. Contrary to previous belief, the effect is not necessarily associated with hypertension and is mediated by a deficit in endothelial nitric oxide. Collectively, the evidence suggests a remarkable cellular diversity of the impact of vascular risk factors on the cerebral vasculature and cognition. Whereas long-term longitudinal epidemiological studies are needed to resolve the temporal relationships between vascular risk factors and cognitive dysfunction, single-cell molecular studies of the vasculature in animal models will provide a fuller mechanistic understanding. This knowledge is critical for developing new preventive, diagnostic, and therapeutic approaches for these devastating diseases of the mind.
Collapse
Affiliation(s)
- Monica M Santisteban
- Feil Family Brain and Mind Research Institute Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
23
|
Brown R, Benveniste H, Black SE, Charpak S, Dichgans M, Joutel A, Nedergaard M, Smith KJ, Zlokovic BV, Wardlaw JM. Understanding the role of the perivascular space in cerebral small vessel disease. Cardiovasc Res 2018; 114:1462-1473. [PMID: 29726891 PMCID: PMC6455920 DOI: 10.1093/cvr/cvy113] [Citation(s) in RCA: 223] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/18/2018] [Accepted: 05/02/2018] [Indexed: 12/17/2022] Open
Abstract
Small vessel diseases (SVDs) are a group of disorders that result from pathological alteration of the small blood vessels in the brain, including the small arteries, capillaries and veins. Of the 35-36 million people that are estimated to suffer from dementia worldwide, up to 65% have an SVD component. Furthermore, SVD causes 20-25% of strokes, worsens outcome after stroke and is a leading cause of disability, cognitive impairment and poor mobility. Yet the underlying cause(s) of SVD are not fully understood. Magnetic resonance imaging has confirmed enlarged perivascular spaces (PVS) as a hallmark feature of SVD. In healthy tissue, these spaces are proposed to form part of a complex brain fluid drainage system which supports interstitial fluid exchange and may also facilitate clearance of waste products from the brain. The pathophysiological signature of PVS and what this infers about their function and interaction with cerebral microcirculation, plus subsequent downstream effects on lesion development in the brain has not been established. Here we discuss the potential of enlarged PVS to be a unique biomarker for SVD and related brain disorders with a vascular component. We propose that widening of PVS suggests presence of peri-vascular cell debris and other waste products that form part of a vicious cycle involving impaired cerebrovascular reactivity, blood-brain barrier dysfunction, perivascular inflammation and ultimately impaired clearance of waste proteins from the interstitial fluid space, leading to accumulation of toxins, hypoxia, and tissue damage. Here, we outline current knowledge, questions and hypotheses regarding understanding the brain fluid dynamics underpinning dementia and stroke through the common denominator of SVD.
Collapse
Affiliation(s)
- Rosalind Brown
- Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor's Building, Edinburgh, UK
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, USA
| | - Sandra E Black
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
- Hurvitz Brain Sciences Program, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Canada
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Serge Charpak
- INSERM U1128, Laboratory of Neurophysiology and New Microscopies, Université Paris Descartes, Paris, France
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Anne Joutel
- Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, Université Paris Diderot-Paris 7, Paris, France
- DHU NeuroVasc, Sorbonne Paris Cité, Paris, France
| | - Maiken Nedergaard
- Section for Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Division of Glia Disease and Therapeutics, Center for Translational Neuromedicine, University of Rochester Medical School, Rochester, USA
| | - Kenneth J Smith
- Department of Neuroinflammation, UCL Institute of Neurology, London, UK
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, Chancellor's Building, Edinburgh, UK
- UK Dementia Research Institute at The University of Edinburgh, Chancellor's Building, Edinburgh, UK
- Row Fogo Centre for Research into Ageing and the Brain, The University of Edinburgh, Chancellor's Building, Edinburgh, UK
| |
Collapse
|
24
|
Pedragosa J, Salas-Perdomo A, Gallizioli M, Cugota R, Miró-Mur F, Briansó F, Justicia C, Pérez-Asensio F, Marquez-Kisinousky L, Urra X, Gieryng A, Kaminska B, Chamorro A, Planas AM. CNS-border associated macrophages respond to acute ischemic stroke attracting granulocytes and promoting vascular leakage. Acta Neuropathol Commun 2018; 6:76. [PMID: 30092836 PMCID: PMC6083589 DOI: 10.1186/s40478-018-0581-6] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/04/2018] [Indexed: 01/16/2023] Open
Abstract
The central nervous system (CNS) contains several types of immune cells located in specific anatomic compartments. Macrophages reside at the CNS borders surrounding the brain vessels, in leptomeningeal spaces and the choroid plexus, where they interact with the vasculature and play immunological surveillance and scavenging functions. We investigated the phenotypic changes and role of these macrophages in response to acute ischemic stroke. Given that CD163 expression is a hallmark of perivascular and meningeal macrophages in the rat and human brain, we isolated CD163+ brain macrophages by fluorescence activated cell sorting. We obtained CD163+ cells from control rats and 16 h following transient middle cerebral artery occlusion, after verifying that infiltration of CD163+ peripheral myeloid cells is negligible at this acute time point. Transcriptome analysis of the sorted CD163+ cells identified ischemia-induced upregulation of the hypoxia inducible factor-1 pathway and induction of genes encoding for extracellular matrix components and leukocyte chemoattractants, amongst others. Using a cell depletion strategy, we found that CNS border-associated macrophages participate in granulocyte recruitment, promote the expression of vascular endothelial growth factor (VEGF), increase the permeability of pial and cortical blood vessels, and contribute to neurological dysfunction in the acute phase of ischemia/reperfusion. We detected VEGF expression surrounding blood vessels and in some CD163+ perivascular macrophages in the brain tissue of ischemic stroke patients deceased one day after stroke onset. These findings show ischemia-induced reprogramming of the gene expression profile of CD163+ macrophages that has a rapid impact on leukocyte chemotaxis and blood-brain barrier integrity, and promotes neurological impairment in the acute phase of stroke.
Collapse
|
25
|
Pires PW, McClain JL, Hayoz SF, Dorrance AM. Mineralocorticoid receptor antagonism prevents obesity-induced cerebral artery remodeling and reduces white matter injury in rats. Microcirculation 2018; 25:e12460. [PMID: 29758591 DOI: 10.1111/micc.12460] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 05/06/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Midlife obesity is a risk factor for dementia development. Obesity has also been linked to hyperaldosteronism, and this can be modeled in rats by high fat (HF) feeding from weaning. Aldosterone, or activation of the mineralocorticoid receptor (MR) causes cerebrovascular injury in lean hypertensive rats. We hypothesized that rats fed a HF diet would show inward middle cerebral artery (MCA) remodeling that could be prevented by MR antagonism. We further proposed that the cerebral artery remodeling would be associated with white mater injury. METHODS Three-week-old male Sprague-Dawley rats were fed a HF diet ± the MR antagonist canrenoic acid (Canr) for 17 weeks. Control rats received normal chow (control NC). MCA structure was assessed by pressure myography. RESULTS The MCAs from HF fed rats had smaller lumens and thicker walls when compared to arteries from control NC rats; Canr prevented the MCA remodeling associated with HF feeding. HF feeding increased the mRNA expression of markers of cell proliferation and vascular inflammation in cerebral arteries and Canr treatment prevented this. White mater injury was increased in the rats fed the HF diet and this was reduced by Canr treatment. The expression of doublecortin, a marker of new and immature neurons was reduced in HF fed rats, and MR antagonism normalized this. CONCLUSIONS These data suggest that HF feeding leads to MR dependent remodeling of the MCA and this is associated with markers of dementia development.
Collapse
Affiliation(s)
- Paulo W Pires
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA.,Department of Pharmacology, Center for Cardiovascular Research, University of Nevada School of Medicine, Reno, NV, USA
| | - Jonathon L McClain
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Sebastian F Hayoz
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA.,Department of Biomedical Sciences, East Tennessee State University, Johnson City, TN, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
26
|
Faraco G, Park L, Anrather J, Iadecola C. Brain perivascular macrophages: characterization and functional roles in health and disease. J Mol Med (Berl) 2017; 95:1143-1152. [PMID: 28782084 DOI: 10.1007/s00109-017-1573-x] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/21/2017] [Accepted: 07/28/2017] [Indexed: 12/12/2022]
Abstract
Perivascular macrophages (PVM) are a distinct population of resident brain macrophages characterized by a close association with the cerebral vasculature. PVM migrate from the yolk sac into the brain early in development and, like microglia, are likely to be a self-renewing cell population that, in the normal state, is not replenished by circulating monocytes. Increasing evidence implicates PVM in several disease processes, ranging from brain infections and immune activation to regulation of the hypothalamic-adrenal axis and neurovascular-neurocognitive dysfunction in the setting of hypertension, Alzheimer disease pathology, or obesity. These effects involve crosstalk between PVM and cerebral endothelial cells, interaction with circulating immune cells, and/or production of reactive oxygen species. Overall, the available evidence supports the idea that PVM are a key component of the brain-resident immune system with broad implications for the pathogenesis of major brain diseases. A better understanding of the biology and pathobiology of PVM may lead to new insights and therapeutic strategies for a wide variety of brain diseases.
Collapse
Affiliation(s)
- Giuseppe Faraco
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 E61st Street, New York, NY, USA.
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 E61st Street, New York, NY, USA
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 E61st Street, New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 E61st Street, New York, NY, USA.
| |
Collapse
|
27
|
Park L, Uekawa K, Garcia-Bonilla L, Koizumi K, Murphy M, Pistik R, Younkin L, Younkin S, Zhou P, Carlson G, Anrather J, Iadecola C. Brain Perivascular Macrophages Initiate the Neurovascular Dysfunction of Alzheimer Aβ Peptides. Circ Res 2017; 121:258-269. [PMID: 28515043 DOI: 10.1161/circresaha.117.311054] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
RATIONALE Increasing evidence indicates that alterations of the cerebral microcirculation may play a role in Alzheimer disease, the leading cause of late-life dementia. The amyloid-β peptide (Aβ), a key pathogenic factor in Alzheimer disease, induces profound alterations in neurovascular regulation through the innate immunity receptor CD36 (cluster of differentiation 36), which, in turn, activates a Nox2-containing NADPH oxidase, leading to cerebrovascular oxidative stress. Brain perivascular macrophages (PVM) located in the perivascular space, a major site of brain Aβ collection and clearance, are juxtaposed to the wall of intracerebral resistance vessels and are a powerful source of reactive oxygen species. OBJECTIVE We tested the hypothesis that PVM are the main source of reactive oxygen species responsible for the cerebrovascular actions of Aβ and that CD36 and Nox2 in PVM are the molecular substrates of the effect. METHODS AND RESULTS Selective depletion of PVM using intracerebroventricular injection of clodronate abrogates the reactive oxygen species production and cerebrovascular dysfunction induced by Aβ applied directly to the cerebral cortex, administered intravascularly, or overproduced in the brain of transgenic mice expressing mutated forms of the amyloid precursor protein (Tg2576 mice). In addition, using bone marrow chimeras, we demonstrate that PVM are the cells expressing CD36 and Nox2 responsible for the dysfunction. Thus, deletion of CD36 or Nox2 from PVM abrogates the deleterious vascular effects of Aβ, whereas wild-type PVM reconstitute the vascular dysfunction in CD36-null mice. CONCLUSIONS The data identify PVM as a previously unrecognized effector of the damaging neurovascular actions of Aβ and unveil a new mechanism by which brain-resident innate immune cells and their receptors may contribute to the pathobiology of Alzheimer disease.
Collapse
Affiliation(s)
- Laibaik Park
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.).
| | - Ken Uekawa
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Lidia Garcia-Bonilla
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Kenzo Koizumi
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Michelle Murphy
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Rose Pistik
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Linda Younkin
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Steven Younkin
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Ping Zhou
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - George Carlson
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Josef Anrather
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Costantino Iadecola
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.).
| |
Collapse
|
28
|
Ungvari Z, Tarantini S, Kirkpatrick AC, Csiszar A, Prodan CI. Cerebral microhemorrhages: mechanisms, consequences, and prevention. Am J Physiol Heart Circ Physiol 2017; 312:H1128-H1143. [PMID: 28314762 PMCID: PMC5495931 DOI: 10.1152/ajpheart.00780.2016] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/22/2017] [Accepted: 03/09/2017] [Indexed: 12/20/2022]
Abstract
The increasing prevalence of multifocal cerebral microhemorrhages (CMHs, also known as "cerebral microbleeds") is a significant, newly recognized problem in the aging population of the Western world. CMHs are associated with rupture of small intracerebral vessels and are thought to progressively impair neuronal function, potentially contributing to cognitive decline, geriatric psychiatric syndromes, and gait disorders. Clinical studies show that aging and hypertension significantly increase prevalence of CMHs. CMHs are also now recognized by the National Institutes of Health as a major factor in Alzheimer's disease pathology. Moreover, the presence of CMHs is an independent risk factor for subsequent larger intracerebral hemorrhages. In this article, we review the epidemiology, detection, risk factors, clinical significance, and pathogenesis of CMHs. The potential age-related cellular mechanisms underlying the development of CMHs are discussed, with a focus on the structural determinants of microvascular fragility, age-related alterations in cerebrovascular adaptation to hypertension, the role of oxidative stress and matrix metalloproteinase activation, and the deleterious effects of arterial stiffening, increased pulse pressure, and impaired myogenic autoregulatory protection on the brain microvasculature. Finally, we examine potential treatments for the prevention of CMHs based on the proposed model of aging- and hypertension-dependent activation of the reactive oxygen species-matrix metalloproteinases axis, and we discuss critical questions to be addressed by future studies.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; .,Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Stefano Tarantini
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.,Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Angelia C Kirkpatrick
- Veterans Affairs Medical Center, Oklahoma City, Oklahoma.,Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and
| | - Anna Csiszar
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.,Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Calin I Prodan
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
29
|
Faraco G, Sugiyama Y, Lane D, Garcia-Bonilla L, Chang H, Santisteban MM, Racchumi G, Murphy M, Van Rooijen N, Anrather J, Iadecola C. Perivascular macrophages mediate the neurovascular and cognitive dysfunction associated with hypertension. J Clin Invest 2016; 126:4674-4689. [PMID: 27841763 DOI: 10.1172/jci86950] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 09/30/2016] [Indexed: 01/05/2023] Open
Abstract
Hypertension is a leading risk factor for dementia, but the mechanisms underlying its damaging effects on the brain are poorly understood. Due to a lack of energy reserves, the brain relies on continuous delivery of blood flow to its active regions in accordance with their dynamic metabolic needs. Hypertension disrupts these vital regulatory mechanisms, leading to the neuronal dysfunction and damage underlying cognitive impairment. Elucidating the cellular bases of these impairments is essential for developing new therapies. Perivascular macrophages (PVMs) represent a distinct population of resident brain macrophages that serves key homeostatic roles but also has the potential to generate large amounts of reactive oxygen species (ROS). Here, we report that PVMs are critical in driving the alterations in neurovascular regulation and attendant cognitive impairment in mouse models of hypertension. This effect was mediated by an increase in blood-brain barrier permeability that allowed angiotensin II to enter the perivascular space and activate angiotensin type 1 receptors in PVMs, leading to production of ROS through the superoxide-producing enzyme NOX2. These findings unveil a pathogenic role of PVMs in the neurovascular and cognitive dysfunction associated with hypertension and identify these cells as a putative therapeutic target for diseases associated with cerebrovascular oxidative stress.
Collapse
|
30
|
Gorelick PB, Counts SE, Nyenhuis D. Vascular cognitive impairment and dementia. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1862:860-8. [PMID: 26704177 PMCID: PMC5232167 DOI: 10.1016/j.bbadis.2015.12.015] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 11/12/2015] [Accepted: 12/14/2015] [Indexed: 01/11/2023]
Abstract
Vascular contributions to cognitive impairment are receiving heightened attention as potentially modifiable factors for dementias of later life. These factors have now been linked not only to vascular cognitive disorders but also Alzheimer's disease. In this chapter we review 3 related topics that address vascular contributions to cognitive impairment: 1. vascular pathogenesis and mechanisms; 2. neuropsychological and neuroimaging phenotypic manifestations of cerebrovascular disease; and 3. prospects for prevention of cognitive impairment of later life based on cardiovascular and stroke risk modification. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
Affiliation(s)
- Philip B Gorelick
- Translational Science & Molecular Medicine, Michigan State University College of Human Medicine, Mercy Health Hauenstein Neurosciences, 220 Cherry Street SE, Grand Rapids, MI 49503, USA.
| | - Scott E Counts
- Translational Science & Molecular Medicine and Family Medicine, Michigan State University College of Human Medicine, Mercy Health Hauenstein Neurosciences, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
| | - David Nyenhuis
- Translational Science & Molecular Medicine, Michigan State University College of Human Medicine, Neuropsychology Program, Mercy Health Hauenstein Neurosciences, 220 Cherry Street SE, Grand Rapids, MI 49503, USA
| |
Collapse
|
31
|
Diaz-Otero JM, Garver H, Fink GD, Jackson WF, Dorrance AM. Aging is associated with changes to the biomechanical properties of the posterior cerebral artery and parenchymal arterioles. Am J Physiol Heart Circ Physiol 2016; 310:H365-75. [PMID: 26637558 PMCID: PMC4796626 DOI: 10.1152/ajpheart.00562.2015] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 12/02/2015] [Indexed: 12/15/2022]
Abstract
Artery remodeling, described as a change in artery structure, may be responsible for the increased risk of cardiovascular disease with aging. Although the risk for stroke is known to increase with age, relatively young animals have been used in most stroke studies. Therefore, more information is needed on how aging alters the biomechanical properties of cerebral arteries. Posterior cerebral arteries (PCAs) and parenchymal arterioles (PAs) are important in controlling brain perfusion. We hypothesized that aged (22-24 mo old) C57bl/6 mice would have stiffer PCAs and PAs than young (3-5 mo old) mice. The biomechanical properties of the PCAs and PAs were assessed by pressure myography. Data are presented as means ± SE of young vs. old. In the PCA, older mice had increased outer (155.6 ± 3.2 vs. 169.9 ± 3.2 μm) and lumen (116.4 ± 3.6 vs. 137.1 ± 4.7 μm) diameters. Wall stress (375.6 ± 35.4 vs. 504.7 ± 60.0 dyn/cm(2)) and artery stiffness (β-coefficient: 5.2 ± 0.3 vs. 7.6 ± 0.9) were also increased. However, wall strain (0.8 ± 0.1 vs. 0.6 ± 0.1) was reduced with age. In the PAs from old mice, wall thickness (3.9 ± 0.3 vs. 5.1 ± 0.2 μm) and area (591.1 ± 95.4 vs. 852.8 ± 100 μm(2)) were increased while stress (758.1 ± 100.0 vs. 587.2 ± 35.1 dyn/cm(2)) was reduced. Aging also increased mean arterial and pulse pressures. We conclude that age-associated remodeling occurs in large cerebral arteries and arterioles and may increase the risk of cerebrovascular disease.
Collapse
Affiliation(s)
- Janice M Diaz-Otero
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Hannah Garver
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Gregory D Fink
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| |
Collapse
|
32
|
Anwar MA, Eid AH. Determination of Vascular Reactivity of Middle Cerebral Arteries from Stroke and Spinal Cord Injury Animal Models Using Pressure Myography. Methods Mol Biol 2016; 1462:611-24. [PMID: 27604741 DOI: 10.1007/978-1-4939-3816-2_33] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Stroke and other neurovascular derangements are main causes of global death. They, along with spinal cord injuries, are responsible for being the principal cause of disability due to neurological and cognitive problems. These problems then lead to a burden on scarce financial resources and societal care facilities as well as have a profound effect on patients' families. The mechanism of action in these debilitating diseases is complex and unclear. An important component of these problems arises from derangement of blood vessels, such as blockage due to clotting/embolism, endothelial dysfunction, and overreactivity to contractile agents, as well as alteration in endothelial permeability. Moreover, the cerebro-vasculature (large vessels and arterioles) is involved in regulating blood flow by facilitating auto-regulatory processes. Moreover, the anterior (middle cerebral artery and the surrounding region) and posterior (basilar artery and its immediate locality) regions of the brain play a significant role in triggering the pathological progression of ischemic stroke particularly due to inflammatory activity and oxidative stress. Interestingly, modifiable and non-modifiable cardiovascular risk factors are responsible for driving ischemic and hemorrhagic stroke and spinal cord injury. There are different stroke animal models to examine the pathophysiology of middle cerebral and basilar arteries. In this context, arterial myography offers an opportunity to determine the etiology of vascular dysfunction in these diseases. Herein, we describe the technique of pressure myography to examine the reactivity of cerebral vessels to contractile and vasodilator agents and a prelude to stroke and spinal cord injury.
Collapse
Affiliation(s)
- Mohammad A Anwar
- Department of Biological & Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Ali H Eid
- Department of Biological & Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar. .,Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, 11-0236, Beirut, 1107-2020, Lebanon.
| |
Collapse
|
33
|
Zhang MZ, Yao B, Wang Y, Yang S, Wang S, Fan X, Harris RC. Inhibition of cyclooxygenase-2 in hematopoietic cells results in salt-sensitive hypertension. J Clin Invest 2015; 125:4281-94. [PMID: 26485285 DOI: 10.1172/jci81550] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 09/03/2015] [Indexed: 01/11/2023] Open
Abstract
Inhibition of prostaglandin (PG) production with either nonselective or selective inhibitors of cyclooxygenase-2 (COX-2) activity can induce or exacerbate salt-sensitive hypertension. This effect has been previously attributed to inhibition of intrinsic renal COX-2 activity and subsequent increase in sodium retention by the kidney. Here, we found that macrophages isolated from kidneys of high-salt-treated WT mice have increased levels of COX-2 and microsomal PGE synthase-1 (mPGES-1). Furthermore, BM transplantation (BMT) from either COX-2-deficient or mPGES-1-deficient mice into WT mice or macrophage-specific deletion of the PGE2 type 4 (EP4) receptor induced salt-sensitive hypertension and increased phosphorylation of the renal sodium chloride cotransporter (NCC). Kidneys from high-salt-treated WT mice transplanted with Cox2-/- BM had increased macrophage and T cell infiltration and increased M1- and Th1-associated markers and cytokines. Skin macrophages from high-salt-treated mice with either genetic or pharmacologic inhibition of the COX-2 pathway expressed decreased M2 markers and VEGF-C production and exhibited aberrant lymphangiogenesis. Together, these studies demonstrate that COX-2-derived PGE2 in hematopoietic cells plays an important role in both kidney and skin in maintaining homeostasis in response to chronically increased dietary salt. Moreover, these results indicate that inhibiting COX-2 expression or activity in hematopoietic cells can result in a predisposition to salt-sensitive hypertension.
Collapse
|
34
|
Pires PW, Jackson WF, Dorrance AM. Regulation of myogenic tone and structure of parenchymal arterioles by hypertension and the mineralocorticoid receptor. Am J Physiol Heart Circ Physiol 2015; 309:H127-36. [PMID: 25910805 DOI: 10.1152/ajpheart.00168.2015] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 04/23/2015] [Indexed: 11/22/2022]
Abstract
Proper perfusion is vital for maintenance of neuronal homeostasis and brain function. Changes in the function and structure of cerebral parenchymal arterioles (PAs) could impair blood flow regulation and increase the risk of cerebrovascular diseases, including dementia and stroke. Hypertension alters the structure and function of large cerebral arteries, but its effects on PAs remain unknown. We hypothesized that hypertension increases myogenic tone and induces inward remodeling in PAs; we further proposed that antihypertensive therapy or mineralocorticoid receptor (MR) blockade would reverse the effects of hypertension. PAs from 18-wk-old stroke-prone spontaneously hypertensive rats (SHRSP) were isolated and cannulated in a pressure myograph. At 50-mmHg intraluminal pressure, PAs from SHRSP showed higher myogenic tone (%tone: 39.1 ± 1.9 vs. 28.7 ± 2.5%, P < 0.01) and smaller resting luminal diameter (34.7 ± 1.9 vs. 46.2 ± 2.4 μm, P < 0.01) than those from normotensive Wistar-Kyoto rats, through a mechanism that seems to require Ca(2+) influx through L-type voltage-gated Ca(2+) channels. PAs from SHRSP showed inward remodeling (luminal diameter at 60 mmHg: 55.2 ± 1.4 vs. 75.7 ± 5.1 μm, P < 0.01) and a paradoxical increase in distensibility and compliance. Treatment of SHRSP for 6 wk with antihypertensive therapy reduced PAs' myogenic tone, increased their resting luminal diameter, and prevented inward remodeling. In contrast, treatment of SHRSP for 6 wk with an MR antagonist did not reduce blood pressure or myogenic tone, but prevented inward remodeling. Thus, while hypertensive remodeling of PAs may involve the MR, myogenic tone seems to be independent of MR activity.
Collapse
Affiliation(s)
- Paulo W Pires
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan; and Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan; and
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan; and
| |
Collapse
|
35
|
Singh P, Gupta S, Sharma B. Melatonin receptor and KATP channel modulation in experimental vascular dementia. Physiol Behav 2015; 142:66-78. [PMID: 25659733 DOI: 10.1016/j.physbeh.2015.02.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 02/03/2015] [Accepted: 02/04/2015] [Indexed: 01/08/2023]
Abstract
Cerebrovascular and cardiovascular diseases are stated as important risk factors of vascular dementia (VaD) and other cognitive disorders. In the central nervous system, melatonin (MT1/MT2) as well as serotonin subtype 2C (5-HT2C) receptors is pharmacologically associated with various neurological disorders. Brain mitochondrial potassium channels have been reported for their role in neuroprotection. This study has been structured to investigate the role of agomelatine, a melatonergic MT1/MT2 agonist and nicorandil, a selective ATP sensitive potassium (KATP) channel opener in renal artery ligation (two-kidney-one-clip: 2K1C) hypertension induced endothelial dysfunction, brain damage and VaD. 2K1C-renovascular hypertension has increased mean arterial blood pressure (MABP), impaired memory (elevated plus maze and Morris water maze), endothelial function, reduced serum nitrite/nitrate and increased brain damage (TTC staining of brain sections). Furthermore, 2K1C animals have shown high levels of oxidative stress in serum (increased thiobarbituric acid reactive species-TBARS with decreased levels of glutathione-GSH, superoxide dismutase-SOD and catalase-CAT), in the aorta (increased aortic superoxide anion) and in the brain (increased TBARS with decreased GSH, SOD and CAT). 2K1C has also induced a significant increase in brain inflammation (myeloperoxidase-MPO levels), acetylcholinesterase activity (AChE) and calcium levels. Impairment in mitochondrial complexes like NADH dehydrogenase (complex-I), succinate dehydrogenase (complex-II) and cytochrome oxidase (complex-IV) was also noted in 2K1C animals. Administration of agomelatine, nicorandil and donepezil significantly attenuated 2K1C-hypertension induced impairments in memory, endothelial function, nitrosative stress, mitochondrial dysfunction, inflammation and brain damage. Therefore, modulators of MT1/MT2 receptors and KATP channels may be considered as potential agents for the management of renovascular hypertension induced VaD.
Collapse
Affiliation(s)
- Prabhat Singh
- CNS and CVS Pharmacology Lab., Department of Pharmacology, School of Pharmacy, Bharat Institute of Technology, Partapur Bypass, Meerut, 250103 Uttar Pradesh, India.
| | - Surbhi Gupta
- CNS and CVS Pharmacology Lab., Department of Pharmacology, School of Pharmacy, Bharat Institute of Technology, Partapur Bypass, Meerut, 250103 Uttar Pradesh, India.
| | - Bhupesh Sharma
- School of Pharmacy, Bharat Institute of Technology, Partapur Bypass, Meerut, 250103 Uttar Pradesh, India; CNS Pharmacology, Conscience Research, Pocket F-233, B, Dilshad Garden, Delhi 110095, India.
| |
Collapse
|
36
|
Murray KN, Parry-Jones AR, Allan SM. Interleukin-1 and acute brain injury. Front Cell Neurosci 2015; 9:18. [PMID: 25705177 PMCID: PMC4319479 DOI: 10.3389/fncel.2015.00018] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 01/12/2015] [Indexed: 01/05/2023] Open
Abstract
Inflammation is the key host-defense response to infection and injury, yet also a major contributor to a diverse range of diseases, both peripheral and central in origin. Brain injury as a result of stroke or trauma is a leading cause of death and disability worldwide, yet there are no effective treatments, resulting in enormous social and economic costs. Increasing evidence, both preclinical and clinical, highlights inflammation as an important factor in stroke, both in determining outcome and as a contributor to risk. A number of inflammatory mediators have been proposed as key targets for intervention to reduce the burden of stroke, several reaching clinical trial, but as yet yielding no success. Many factors could explain these failures, including the lack of robust preclinical evidence and poorly designed clinical trials, in addition to the complex nature of the clinical condition. Lack of consideration in preclinical studies of associated co-morbidities prevalent in the clinical stroke population is now seen as an important omission in previous work. These co-morbidities (atherosclerosis, hypertension, diabetes, infection) have a strong inflammatory component, supporting the need for greater understanding of how inflammation contributes to acute brain injury. Interleukin (IL)-1 is the prototypical pro-inflammatory cytokine, first identified many years ago as the endogenous pyrogen. Research over the last 20 years or so reveals that IL-1 is an important mediator of neuronal injury and blocking the actions of IL-1 is beneficial in a number of experimental models of brain damage. Mechanisms underlying the actions of IL-1 in brain injury remain unclear, though increasing evidence indicates the cerebrovasculature as a key target. Recent literature supporting this and other aspects of how IL-1 and systemic inflammation in general contribute to acute brain injury are discussed in this review.
Collapse
Affiliation(s)
- Katie N Murray
- Faculty of Life Sciences, University of Manchester Manchester, UK
| | | | - Stuart M Allan
- Faculty of Life Sciences, University of Manchester Manchester, UK
| |
Collapse
|
37
|
Onetti Y, Dantas AP, Pérez B, Cugota R, Chamorro A, Planas AM, Vila E, Jiménez-Altayó F. Middle cerebral artery remodeling following transient brain ischemia is linked to early postischemic hyperemia: a target of uric acid treatment. Am J Physiol Heart Circ Physiol 2015; 308:H862-74. [PMID: 25637543 DOI: 10.1152/ajpheart.00001.2015] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 01/28/2015] [Indexed: 12/26/2022]
Abstract
Ischemia impairs blood supply to the brain, and reperfusion is important to restore cerebral blood flow (CBF) and rescue neurons from cell death. However, reperfusion can induce CBF values exceeding the basal values before ischemia. This hyperemic effect has been associated with a worse ischemic brain damage, albeit the mechanisms that contribute to infarct expansion are not clear. In this study, we investigated the influence of early postischemic hyperemia on brain damage and middle cerebral artery (MCA) properties and the effect of treatment with the endogenous antioxidant uric acid (UA). The MCA was occluded for 90 min followed by 24 h reperfusion in adult male Sprague-Dawley rats. Cortical CBF increases at reperfusion beyond 20% of basal values were taken as indicative of hyperemia. UA (16 mg/kg) or vehicle (Locke's buffer) was administered intravenously 135 min after MCA occlusion. Hyperemic compared with nonhyperemic rats showed MCA wall thickening (sham: 22.4 ± 0.8 μm; nonhyperemic: 23.1 ± 1.2 μm; hyperemic: 27.8 ± 0.9 at 60 mmHg; P < 0.001, hyperemic vs. sham) involving adventitial cell proliferation, increased oxidative stress, and interleukin-18, and more severe brain damage. Thus MCA remodeling after ischemia-reperfusion takes place under vascular oxidative and inflammatory stress conditions linked to hyperemia. UA administration attenuated MCA wall thickening, induced passive lumen expansion, and reduced brain damage in hyperemic rats, although it did not increase brain UA concentration. We conclude that hyperemia at reperfusion following brain ischemia induces vascular damage that can be attenuated by administration of the endogenous antioxidant UA.
Collapse
Affiliation(s)
- Yara Onetti
- Facultat de Medicina, Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Ana P Dantas
- Institut Clínic del Tòrax, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Belén Pérez
- Facultat de Medicina, Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Roger Cugota
- Facultat de Medicina, Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Angel Chamorro
- Functional Unit of Cerebrovascular Diseases, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; and
| | - Anna M Planas
- Departament d'Isquèmia Cerebral i Neurodegeneració, Institut de Recerca Biomèdica, Consejo Superior de Investigaciones Científicas, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Elisabet Vila
- Facultat de Medicina, Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Francesc Jiménez-Altayó
- Facultat de Medicina, Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain;
| |
Collapse
|
38
|
Abstract
A large number of investigations have demonstrated the participation of the immune system in the pathogenesis of hypertension. Studies focusing on macrophages and Toll-like receptors have documented involvement of the innate immunity. The requirements of antigen presentation and co-stimulation, the critical importance of T cell-driven inflammation, and the demonstration, in specific conditions, of agonistic antibodies directed to angiotensin II type 1 receptors and adrenergic receptors support the role of acquired immunity. Experimental findings support the concept that the balance between T cell-induced inflammation and T cell suppressor responses is critical for the regulation of blood pressure levels. Expression of neoantigens in response to inflammation, as well as surfacing of intracellular immunogenic proteins, such as heat shock proteins, could be responsible for autoimmune reactivity in the kidney, arteries, and central nervous system. Persisting, low-grade inflammation in these target organs may lead to impaired pressure natriuresis, an increase in sympathetic activity, and vascular endothelial dysfunction that may be the cause of chronic elevation of blood pressure in essential hypertension.
Collapse
Affiliation(s)
- Bernardo Rodríguez-Iturbe
- Hospital Universitario y Universidad del Zulia, Maracaibo, Venezuela; Instituto Venezolano de Investigaciones Científicas-Zulia, Maracaibo, Venezuela;
| | - Héctor Pons
- Hospital Universitario y Universidad del Zulia, Maracaibo, Venezuela
| | - Yasmir Quiroz
- Instituto Venezolano de Investigaciones Científicas-Zulia, Maracaibo, Venezuela
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado, Denver, Colorado
| |
Collapse
|
39
|
Pires PW, Girgla SS, Moreno G, McClain JL, Dorrance AM. Tumor necrosis factor-α inhibition attenuates middle cerebral artery remodeling but increases cerebral ischemic damage in hypertensive rats. Am J Physiol Heart Circ Physiol 2014; 307:H658-69. [PMID: 25015967 DOI: 10.1152/ajpheart.00018.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hypertension causes vascular inflammation evidenced by an increase in perivascular macrophages and proinflammatory cytokines in the arterial wall. Perivascular macrophage depletion reduced tumor necrosis factor (TNF)-α expression in cerebral arteries of hypertensive rats and attenuated inward remodeling, suggesting that TNF-α might play a role in the remodeling process. We hypothesized that TNF-α inhibition would improve middle cerebral artery (MCA) structure and reduce damage after cerebral ischemia in hypertensive rats. Six-week-old male stroke-prone spontaneously hypertensive rats (SHRSP) were treated with the TNF-α inhibitor etanercept (ETN; 1.25 mg·kg(-1)·day(-1) ip daily) or PBS (equivolume) for 6 wk. The myogenic tone generation, postischemic dilation, and passive structure of MCAs were assessed by pressure myography. Cerebral ischemia was induced by MCA occlusion (MCAO). Myogenic tone was unchanged, but MCAs from SHRSP + ETN had larger passive lumen diameter and reduced wall thickness and wall-to-lumen ratio. Cerebral infarct size was increased in SHRSP + ETN after transient MCAO, despite an improvement in dilation of nonischemic MCA. The increase in infarct size was linked to a reduction in the number of microglia in the infarct core and upregulation of markers of classical macrophage/microglia polarization. There was no difference in infarct size after permanent MCAO or when untreated SHRSP subjected to transient MCAO were given ETN at reperfusion. Our data suggests that TNF-α inhibition attenuates hypertensive MCA remodeling but exacerbates cerebral damage following ischemia/reperfusion injury likely due to inhibition of the innate immune response of the brain.
Collapse
Affiliation(s)
- Paulo W Pires
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Saavia S Girgla
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Guillermo Moreno
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Jonathon L McClain
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| |
Collapse
|
40
|
McClain JL, Dorrance AM. Temporary mineralocorticoid receptor antagonism during the development of hypertension improves cerebral artery dilation. Exp Biol Med (Maywood) 2014; 239:619-27. [PMID: 24625441 DOI: 10.1177/1535370214522586] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Hypertension causes cerebral artery remodeling and increases the risk of stroke. Renin angiotensin system blockade during the development of hypertension has therapeutic effects even after treatment withdrawal. Mineralocorticoid receptor (MR) activation has been implicated in artery remodeling and impaired endothelial function. The possibility that there is a critical therapeutic window for MR antagonism has not been investigated. We hypothesized that temporary MR antagonism while hypertension develops would improve middle cerebral artery (MCA) structure and function in stroke-prone spontaneously hypertensive rats (SHRSP), even after treatment withdrawal. Six-week-old SHRSP were treated with spironolactone (25 mg/kg/day) from 6 to 12 weeks and when aged to 18 weeks, these rats were compared to age-matched untreated SHRSP. Surprisingly, temporary spironolactone treatment reduced the MCA outer and lumen diameter but had no effect on the wall thickness. Temporary spironolactone treatment improved nitric oxide and endothelium-derived hyperpolarizing factor mediated dilation but had no effect on blood pressure. Spironolactone treatment caused a very small reduction in the damage caused by permanent focal cerebral ischemia. These results suggest that temporary MR antagonism during the development of hypertension has divergent effects on the MCA, in that it causes a potentially detrimental reduction in the lumen diameter while improving vasodilation.
Collapse
Affiliation(s)
- Jonathon L McClain
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | | |
Collapse
|